51
|
Wang SV, Kulldorff M, Poor S, Rice DS, Banks A, Li N, Lii J, Gagne JJ. Screening Medications for Association with Progression to Wet Age-Related Macular Degeneration. Ophthalmology 2020; 128:248-255. [PMID: 32777229 DOI: 10.1016/j.ophtha.2020.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 11/27/2022] Open
Abstract
PURPOSE There is an urgent need for treatments that prevent or delay development to advanced age-related macular degeneration (AMD). Drugs already on the market for other conditions could affect progression to neovascular AMD (nAMD). If identified, these drugs could provide insights for drug development targets. The objective of this study was to use a novel data mining method that can simultaneously evaluate thousands of correlated hypotheses, while adjusting for multiple testing, to screen for associations between drugs and delayed progression to nAMD. DESIGN We applied a nested case-control study to administrative insurance claims data to identify cases with nAMD and risk-set sampled controls that were 1:4 variable ratio matched on age, gender, and recent healthcare use. PARTICIPANTS The study population included cases with nAMD and risk set matched controls. METHODS We used a tree-based scanning method to evaluate associations between hierarchical classifications of drugs that patients were exposed to within 6 months, 7 to 24 months, or ever before their index date. The index date was the date of first nAMD diagnosis in cases. Risk-set sampled controls were assigned the same index date as the case to which they were matched. The study was implemented using Medicare data from New Jersey and Pennsylvania, and national data from IBM MarketScan Research Database. We set an a priori threshold for statistical alerting at P ≤ 0.01 and focused on associations with large magnitude (relative risks ≥ 2.0). MAIN OUTCOME MEASURES Progression to nAMD. RESULTS Of approximately 4000 generic drugs and drug classes evaluated, the method detected 19 distinct drug exposures with statistically significant, large relative risks indicating that cases were less frequently exposed than controls. These included (1) drugs with prior evidence for a causal relationship (e.g., megestrol); (2) drugs without prior evidence for a causal relationship, but potentially worth further exploration (e.g., donepezil, epoetin alfa); (3) drugs with alternative biologic explanations for the association (e.g., sevelamer); and (4) drugs that may have resulted in statistical alerts due to their correlation with drugs that alerted for other reasons. CONCLUSIONS This exploratory drug-screening study identified several potential targets for follow-up studies to further evaluate and determine if they may prevent or delay progression to advanced AMD.
Collapse
Affiliation(s)
- Shirley V Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Martin Kulldorff
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Stephen Poor
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Dennis S Rice
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Angela Banks
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Ning Li
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Joyce Lii
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joshua J Gagne
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
52
|
Xu W, Su BJ, Shen XN, Bi YL, Tan CC, Li JQ, Cao XP, Dong Q, Tan L, Yu JT. Plasma sex hormone-binding globulin predicts neurodegeneration and clinical progression in prodromal Alzheimer's disease. Aging (Albany NY) 2020; 12:14528-14541. [PMID: 32699184 PMCID: PMC7425468 DOI: 10.18632/aging.103497] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/01/2020] [Indexed: 11/25/2022]
Abstract
It was unclear whether sex hormone-binding globulin (SHBG) was a circulating biomarker of Alzheimer’s disease (AD). We tested the cross-sectional relationships between plasma SHBG and cerebrospinal fluid (CSF) AD biomarkers in 707 non-demented adults. Next, the influences of plasma SHBG on dynamic changes of CSF Aβ42, hippocampus volume, brain metabolism, and cognition were explored in 448 non-demented adults from the Alzheimer’s disease Neuroimaging Initiative (ADNI). Finally, the predictive and diagnostic values of plasma SHBG in AD were explored. A positive correlation was found between SHBG levels in plasma and CSF. Individuals with higher plasma SHBG levels had lower CSF Aβ42 (p < 0.005), after adjusting for age, gender, education, APOE4 allele, and cognitive scores. Though no significant difference of plasma SHBG was observed between mild AD dementia and healthy normal, plasma SHBG could contribute to accelerated rates of CSF Aβ42 decrease (p < 0.0005), decline in brain metabolism (p < 0.05), and hippocampus atrophy (p < 0.01), cognitive decline (p < 0.01), as well as higher risk of AD dementia (p < 0.05). These findings indicated plasma SHBG could be a prodromal biomarker to predict disease progression in AD.
Collapse
Affiliation(s)
- Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Bing-Jie Su
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan-Lin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jie-Qiong Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | | | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
53
|
Wu Z, Wu H, Sun S, Wu H, Shi W, Song J, Liu J, Zhang Y, Bian F, Jia P, Hou Y. Progesterone attenuates Aβ25–35-induced neuronal toxicity by activating the Ras signalling pathway through progesterone receptor membrane component 1. Life Sci 2020; 253:117360. [DOI: 10.1016/j.lfs.2020.117360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 12/23/2022]
|
54
|
Joseph V, Laouafa S, Marcouiller F, Roussel D, Pialoux V, Bairam A. Progesterone decreases apnoea and reduces oxidative stress induced by chronic intermittent hypoxia in ovariectomized female rats. Exp Physiol 2020; 105:1025-1034. [PMID: 32196792 DOI: 10.1113/ep088430] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does progesterone reduce the effect of chronic intermittent hypoxia (CIH) on arterial blood pressure, respiratory control and oxidative stress in the central nervous system in ovariectomized rats? What is the main finding and its importance? Progesterone does not prevent the elevation of arterial blood pressure in rats exposed to CIH, but normalizes respiratory control, and reduces cerebral oxidative stress. This study draws focus to a potential role of progesterone and the consequences of sleep apnoea in menopausal women. ABSTRACT We tested the hypothesis that progesterone (Prog) reduces the effect of chronic intermittent hypoxia (CIH) on arterial blood pressure, respiratory chemoreflexes and oxidative stress in the central nervous system. Ovariectomized female rats were implanted with osmotic pumps delivering vehicle (Veh) or Prog (4 mg kg-1 day-1 ). Two weeks following the surgery, rats were exposed to room air (Air) or CIH (7 days, 10% O2 , 10 cycles h-1 , 8 h day-1 ). We studied three groups: Veh-Air, Veh-CIH and Prog-CIH. After the CIH exposures, we measured the mean arterial pressure (MAP; tail cuff) and assessed the frequency of apnoeas at rest and ventilatory responses to hypoxia and hypercapnia (whole body plethysmography). The activities of the pro-oxidant enzyme NADPH oxidase (NOX) and antioxidant enzymes superoxide dismutase (SOD; in mitochondrial and cytosolic fractions) and glutathione peroxidase (GPx), as well as the concentration of malondialdehyde (MDA), a marker of lipid peroxidation, were measured in brain cortex and brainstem samples. CIH exposure increased the MAP, the frequency of apnoeas, and the respiratory frequency response to hypoxia and hypercapnia. Prog did not prevent the CIH-induced elevation in MAP, but it reduced the CIH-induced frequency of apnoeas and increased hypoxic and hypercapnic ventilatory responses. In the brain cortex, CIH increased NOX activity, and decreased the cytosolic and mitochondrial SOD activities. These effects were prevented by Prog. NOX activity was increased by CIH in the brainstem, and this was also blocked by Prog. The study draws focus to the links between ovarian hormones and the consequences of sleep apnoea in women.
Collapse
Affiliation(s)
- Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Sofien Laouafa
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada.,University of Lyon, Université Claude Bernard Lyon 1, CNRS, ENTPE, UMR5023 LEHNA, Villeurbanne, F-69622, France
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Damien Roussel
- University of Lyon, Université Claude Bernard Lyon 1, CNRS, ENTPE, UMR5023 LEHNA, Villeurbanne, F-69622, France
| | - Vincent Pialoux
- University of Lyon, Université Claude Bernard Lyon 1, LIBM EA 7424, Villeurbanne, 69622, France.,Institut Universitaire de France, Paris, France
| | - Aida Bairam
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
55
|
A U-shaped Association of Breastfeeding Duration with Cognitive Impairment in Chinese Postmenopausal Women. Sci Rep 2020; 10:6584. [PMID: 32313098 PMCID: PMC7170904 DOI: 10.1038/s41598-020-63599-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/01/2020] [Indexed: 11/21/2022] Open
Abstract
Breastfeeding is related to maternal health. However, the association of women’s breastfeeding duration with cognitive function in their later life is limited and inconsistent. The aim of this study was to accurately evaluate the association in Chinese postmenopausal women. We analyzed the data from Zhejiang Ageing and Health Cohort Study including 5487 postmenopausal women. Cognitive impairment was assessed via the Mini-Mental State Examination. Data on breastfeeding duration was collected in the reproductive history section within the questionnaire. Generalized additive models (GAMs) and logistic regression models, controlled for an extensive range of potential confounders, were generated to examine the associations. A U-shaped association was identified between breastfeeding duration and cognitive impairment based on GAM. The nadir with lowest odds of cognitive impairment was ascertained by quadratic model as 12 months. The logistic models showed that compared with women breastfeeding 12 months per child, the fully adjusted odds ratios (ORs) were 1.50 (95% Confidence Interval (CI): 1.20–1.88), 1.58 (95% CI: 1.29–1.93), 1.33 (95% CI: 1.06–1.68), 2.08 (95% CI: 1.64–2.65) for those averagely breastfeeding <6, 6-<12,>12–18,>18 months, respectively. Furthermore, we did not observe significant effect modification of the association. Future longitudinal studies are needed to confirm the association.
Collapse
|
56
|
Gurvich C, Warren AM, Worsley R, Hudaib AR, Thomas N, Kulkarni J. Effects of Oral Contraceptive Androgenicity on Visuospatial and Social-Emotional Cognition: A Prospective Observational Trial. Brain Sci 2020; 10:brainsci10040194. [PMID: 32218215 PMCID: PMC7226060 DOI: 10.3390/brainsci10040194] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/14/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022] Open
Abstract
Oral contraceptives (OCs) containing estrogen and progesterone analogues are widely used amongst reproductive-aged women, but their neurocognitive impact is poorly understood. Preliminary studies suggest that OCs improve verbal memory and that OCs with greater androgenic activity may improve visuospatial ability. We sought to explore the cognitive impact of OCs by assessing performance of OC users at different stages of the OC cycle, and comparing this performance between users of different OC formulations according to known androgenic activity. We conducted a prospective, observational trial of OC users, evaluating cognitive performance with CogState software on two occasions: days 7-10 of active hormonal pill phase, and days 3-5 of the inactive pill phase (coinciding with the withdrawal bleed resembling menstruation). Thirty-five OC users (18 taking androgenic formulations, 17 taking anti-androgenic) were assessed. Analysis by androgenic activity showed superior performance by users of androgenic OCs, as compared to anti-androgenic OCs, in visuospatial ability and facial affect discrimination tasks. A growing understanding of cognitive effects of OC progestin androgenicity may have implications in choice of OC formulation for individuals and in future OC development.
Collapse
|
57
|
Ungard RG, Zhu YF, Yang S, Nakhla P, Parzei N, Zhu KL, Singh G. Response to pregabalin and progesterone differs in male and female rat models of neuropathic and cancer pain. CANADIAN JOURNAL OF PAIN-REVUE CANADIENNE DE LA DOULEUR 2020; 4:39-58. [PMID: 33987485 PMCID: PMC7951160 DOI: 10.1080/24740527.2020.1724776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: Cancer pain involves nervous system damage and pathological neurogenesis. Neuropathic pain arises from damage to the nervous system and is driven by ectopic signaling. Both progesterone and pregabalin are neuroprotective in animal models, and there is evidence that both drugs bind to and inhibit voltage-gated calcium channels. Aims: This study was designed to characterize the effects of progesterone and pregabalin in preclinical models of cancer and neuropathic pain in both sexes. Methods: We measured peripheral sensory signaling by intracellular in vivo electrophysiology and behavioral indicators of pain in rat models of cancer-induced bone pain and neuropathic pain. Results: Female but not male models of cancer pain showed a behavioral response to treatment and pregabalin reduced excitability in C and A high-threshold but not low-threshold sensory neurons of both sexes. Male models of neuropathic pain treated with pregabalin demonstrated higher signaling thresholds only in A high-threshold neurons, and behavioral data indicated a clear recovery to baseline mechanical withdrawal thresholds in all treatment groups. Female rat treatment groups did not show excitability changes in sensory neurons, but all demonstrated higher mechanical withdrawal thresholds than vehicle-treated females, although not to baseline levels. Athymic female rat models of neuropathic pain showed no behavioral or electrophysiological responses to treatment. Conclusions: Both pregabalin and progesterone showed evidence of efficacy in male models of neuropathic pain. These results add to the evidence demonstrating differential effects of treatments for pain in male and female animals and widely differing responses in models of cancer and neuropathic pain.
Collapse
Affiliation(s)
- Robert G Ungard
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Yong Fang Zhu
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sarah Yang
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Peter Nakhla
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Natalka Parzei
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kan Lun Zhu
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gurmit Singh
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
58
|
Tsutsui K, Haraguchi S. Neuroprotective actions of cerebellar and pineal allopregnanolone on Purkinje cells. FASEB Bioadv 2020; 2:149-159. [PMID: 32161904 PMCID: PMC7059624 DOI: 10.1096/fba.2019-00055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/12/2019] [Accepted: 01/08/2020] [Indexed: 11/23/2022] Open
Abstract
The brain produces steroids de novo from cholesterol, so‐called “neurosteroids.” The Purkinje cell, a cerebellar neuron, was discovered as a major site of the biosynthesis of neurosteroids including sex steroids, such as progesterone, from cholesterol in the brain. Allopregnanolone, a progesterone metabolite, is also synthesized in the cerebellum and acts on the Purkinje cell to prevent cell death of this neuron. Recently, the pineal gland was discovered as an important site of the biosynthesis of neurosteroids. Allopregnanolone, a major pineal neurosteroid, acts on the Purkinje cell for the survival of this neuron by suppressing the expression of caspase‐3, a crucial mediator of apoptosis. This review summarizes the discovery of cerebellar and pineal allopregnanolone and its neuroprotective action on Purkinje cells.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences Department of Biology Waseda University Center for Medical Life Science of Waseda University Tokyo Japan
| | - Shogo Haraguchi
- Laboratory of Integrative Brain Sciences Department of Biology Waseda University Center for Medical Life Science of Waseda University Tokyo Japan.,Department of Biochemistry Showa University School of Medicine Tokyo Japan
| |
Collapse
|
59
|
McGarry A, McDermott MP, Kieburtz K, Peng J, Cudkowicz M. Baseline Variables Associated with Functional Decline in 2CARE, A Randomized Clinical Trial in Huntington's Disease. J Huntingtons Dis 2020; 9:47-58. [PMID: 31985471 DOI: 10.3233/jhd-190391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Despite the clearly recognized progressive functional decline of Huntington's disease (HD), detailed investigations of factors associated with the rate of functional progression are limited. OBJECTIVE Understanding factors associated with functional decline through examination of existing HD clinical databases may improve efforts to mitigate it. METHODS We analyzed data from 2CARE, a randomized clinical trial with up to 5 years of follow-up, to assess potential risk factors for more rapid functional decline in HD. RESULTS Variables associated with faster functional decline included worse motor performance, worse cognitive test scores, female sex, lower weight and body mass index, and a higher CAG repeat length, especially in younger people. CONCLUSION While our data are limited to the structured environment and homogeneity of a clinical trial, attention to several of the identified risk factors may be useful towards managing functional decline over time. The observation that women progress faster than men, while potentially confounded by an association between sex and weight, deserves further study.
Collapse
Affiliation(s)
- Andrew McGarry
- Cooper University Healthcare at Rowan University, Camden, NJ, USA
| | | | | | - Jing Peng
- The Ohio State University, Columbus, OH, USA
| | | | | |
Collapse
|
60
|
Marquis JM, Lettenberger SE, Kelm-Nelson CA. Early-onset Parkinsonian behaviors in female Pink1-/- rats. Behav Brain Res 2020; 377:112175. [PMID: 31542395 PMCID: PMC6824965 DOI: 10.1016/j.bbr.2019.112175] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/01/2019] [Accepted: 08/22/2019] [Indexed: 12/26/2022]
Abstract
Parkinson disease (PD) is a progressive, neurological disease that affects millions of individuals worldwide. Although instability, rigidity, tremor, and bradykinesia are considered hallmark motor signs of the disease, these are not apparent until mid-to-late stage. In addition to limb motor impairment, individuals with PD also exhibit early-onset speech dysfunction and reduced vocal intelligibility as well as anhedonia and anxiety. Many of these clinical signs vary according to sex in humans with PD. In this study, a translational genetic rat model of early-onset PD (Pink1-/-) was used to address significant gaps in knowledge concerning sex-specific characteristics of limb sensorimotor deficits, vocal motor dysfunction, and changes in affective state. Traditional behavioral tests of limb function, ultrasonic vocalization, anxiety, and anhedonia in the Pink1-/- female rat and wildtype controls were used to test the hypothesis that behavioral performance would significantly differ between genotypes, and that these differences would increase with disease progression (age of the rat). Results demonstrate that Pink1-/- female rats do not exhibit limb sensorimotor deficits but do have significantly reduced intensity (loudness) of vocalizations, and present with anhedonia and anxiety by 8 months of age. Consistent with an early-disease model, Pink1-/- female rats do not exhibit significant decreases in nigrostriatal catecholamines/metabolites, as measured by HPLC. These results are significant in expanding knowledge of early-onset deficits in the female Pink1-/- genetic rat model of PD.
Collapse
Affiliation(s)
- Julia M Marquis
- Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, USA; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA.
| | - Samantha E Lettenberger
- Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, USA; Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, USA.
| | - Cynthia A Kelm-Nelson
- Department of Surgery, Division of Otolaryngology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
61
|
Yare K, Woodward M. Hormone Therapy and Effects on Sporadic Alzheimer’s Disease in Postmenopausal Women: Importance of Nomenclature. J Alzheimers Dis 2020; 73:23-37. [DOI: 10.3233/jad-190896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Katrine Yare
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| | - Michael Woodward
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| |
Collapse
|
62
|
Zhang P, Chen JS, Li QY, Sheng LX, Gao YX, Lu BZ, Zhu WB, Zhan XY, Li Y, Yuan ZB, Xu G, Qiu BT, Yan M, Guo CX, Wang YQ, Huang YJ, Zhang JX, Liu FY, Tang ZW, Lin SZ, Cooper DN, Yang HM, Wang J, Gao YQ, Yin W, Zhang GJ, Yan GM. Neuroprotectants attenuate hypobaric hypoxia-induced brain injuries in cynomolgus monkeys. Zool Res 2020; 41:3-19. [PMID: 31840949 PMCID: PMC6956719 DOI: 10.24272/j.issn.2095-8137.2020.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hypobaric hypoxia (HH) exposure can cause serious brain injury as well as life-threatening cerebral edema in severe cases. Previous studies on the mechanisms of HH-induced brain injury have been conducted primarily using non-primate animal models that are genetically distant to humans, thus hindering the development of disease treatment. Here, we report that cynomolgus monkeys (Macacafascicularis) exposed to acute HH developed human-like HH syndrome involving severe brain injury and abnormal behavior. Transcriptome profiling of white blood cells and brain tissue from monkeys exposed to increasing altitude revealed the central role of the HIF-1 and other novel signaling pathways, such as the vitamin D receptor (VDR) signaling pathway, in co-regulating HH-induced inflammation processes. We also observed profound transcriptomic alterations in brains after exposure to acute HH, including the activation of angiogenesis and impairment of aerobic respiration and protein folding processes, which likely underlie the pathological effects of HH-induced brain injury. Administration of progesterone (PROG) and steroid neuroprotectant 5α-androst-3β,5,6β-triol (TRIOL) significantly attenuated brain injuries and rescued the transcriptomic changes induced by acute HH. Functional investigation of the affected genes suggested that these two neuroprotectants protect the brain by targeting different pathways, with PROG enhancing erythropoiesis and TRIOL suppressing glutamate-induced excitotoxicity. Thus, this study advances our understanding of the pathology induced by acute HH and provides potential compounds for the development of neuroprotectant drugs for therapeutic treatment.
Collapse
Affiliation(s)
- Pei Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,BGI-Shenzhen, Shenzhen, Guangdong 518083, China.,Section for Ecology and Evolution, Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Jie-Si Chen
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Qi-Ye Li
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Long-Xiang Sheng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yi-Xing Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, China.,Key Laboratory of High Altitude Medicine of People's Liberation Army, Chongqing 400038, China.,Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing 400038, China
| | - Bing-Zheng Lu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Wen-Bo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | | | - Yuan Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zhi-Bing Yuan
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, China.,Key Laboratory of High Altitude Medicine of People's Liberation Army, Chongqing 400038, China.,Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing 400038, China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, China.,Key Laboratory of High Altitude Medicine of People's Liberation Army, Chongqing 400038, China.,Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing 400038, China
| | - Bi-Tao Qiu
- Section for Ecology and Evolution, Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Min Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | | | - You-Qiong Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yi-Jun Huang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Jing-Xia Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Fu-Yu Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, China.,Key Laboratory of High Altitude Medicine of People's Liberation Army, Chongqing 400038, China.,Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing 400038, China
| | - Zhong-Wei Tang
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, China.,Key Laboratory of High Altitude Medicine of People's Liberation Army, Chongqing 400038, China.,Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing 400038, China
| | - Sui-Zhen Lin
- Guangzhou Cellprotek Pharmaceutical Co. Ltd., Guangzhou, Guangdong 510663, China
| | - David N. Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Huan-Ming Yang
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang 310058, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, China.,Key Laboratory of High Altitude Medicine of People's Liberation Army, Chongqing 400038, China.,Key Laboratory of High Altitude Environmental Medicine, Third Military Medical University, Ministry of Education, Chongqing 400038, China. E-mail:
| | - Wei Yin
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China. E-mail:
| | - Guo-Jie Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Section for Ecology and Evolution, Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,China National Genebank, BGI-Shenzhen, Shenzhen, Guangdong 518120, China. E-mail:
| | - Guang-Mei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China. E-mail:
| |
Collapse
|
63
|
Jarras H, Bourque M, Poirier AA, Morissette M, Coulombe K, Di Paolo T, Soulet D. Neuroprotection and immunomodulation of progesterone in the gut of a mouse model of Parkinson's disease. J Neuroendocrinol 2020; 32:e12782. [PMID: 31430407 DOI: 10.1111/jne.12782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
Gastrointestinal symptoms appear in Parkinson's disease patients many years before motor symptoms, suggesting the implication of dopaminergic neurones of the gut myenteric plexus. Inflammation is also known to be increased in PD. We previously reported neuroprotection with progesterone in the brain of mice lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and hypothesised that it also has neuroprotective and immunomodulatory activities in the gut. To test this hypothesis, we investigated progesterone administered to adult male C57BL/6 mice for 10 days and treated with MPTP on day 5. In an additional experiment, progesterone was administered for 5 days following MPTP treatment. Ilea were collected on day 10 of treatment and microdissected to isolate the myenteric plexus. Dopaminergic neurones were reduced by approximately 60% and pro-inflammatory macrophages were increased by approximately 50% in MPTP mice compared to intact controls. These changes were completely prevented by progesterone administered before and after MPTP treatment and were normalised by 8 mg kg-1 progesterone administered after MPTP. In the brain of MPTP mice, brain-derived neurotrophic peptide (BDNF) and glial fibrillary acidic protein (GFAP) were associated with progesterone neuroprotection. In the myenteric plexus, increased BDNF levels compared to controls were measured in MPTP mice treated with 8 mg kg-1 progesterone started post MPTP, whereas GFAP levels remained unchanged. In conclusion, the results obtained in the present study show neuroprotective and anti-inflammatory effects of progesterone in the myenteric plexus of MPTP mice that are similar to our previous findings in the brain. Progesterone is non-feminising and could be used for both men and women in the pre-symptomatic stages of the disease.
Collapse
Affiliation(s)
- Hend Jarras
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Mélanie Bourque
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Andrée-Anne Poirier
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Katherine Coulombe
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| | - Denis Soulet
- Axe Neurosciences, Centre de Recherche du CHU de Québec (Pavillon CHUL), Quebec, Canada
- Faculty of Pharmacy, Laval University, Quebec, Canada
| |
Collapse
|
64
|
Gurvich C, Thomas N, Kulkarni J. Sex differences in cognition and aging and the influence of sex hormones. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:103-115. [PMID: 33008519 DOI: 10.1016/b978-0-444-64123-6.00008-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Sex differences in cognitive functioning have been consistently reported in some cognitive tasks, with varying effect sizes. The most consistent findings in healthy adults are sex differences in the areas of mental rotation and aspects of attention and verbal memory. Sex differences in the vulnerability and manifestation of several psychiatric and neurologic diseases that involve cognitive disruption provide strong justification to continue investigating the social and biologic influences that underpin sex differences in cognitive functioning across health and disease. The biologic influences are thought to include genetic and epigenetic factors, sex chromosomes, and sex hormones. Sex steroid hormones that regulate reproductive function have multiple effects on the development, maintenance, and function of the brain, including significant effects on cognitive functioning. The aim of the current chapter is to provide a theoretical review of sex differences across different cognitive domains in adulthood and aging, as well as provide an overview on the role of sex hormones in cognitive function and cognitive decline.
Collapse
Affiliation(s)
- Caroline Gurvich
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and Alfred Hospital, Melbourne, VIC, Australia.
| | - Natalie Thomas
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and Alfred Hospital, Melbourne, VIC, Australia
| | - Jayashri Kulkarni
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
65
|
Chung YS, Poppe A, Novotny S, Epperson CN, Kober H, Granger DA, Blumberg HP, Ochsner K, Gross JJ, Pearlson G, Stevens MC. A preliminary study of association between adolescent estradiol level and dorsolateral prefrontal cortex activity during emotion regulation. Psychoneuroendocrinology 2019; 109:104398. [PMID: 31394491 PMCID: PMC6842698 DOI: 10.1016/j.psyneuen.2019.104398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 12/19/2022]
Abstract
Non-human primate models have been useful in clarifying estradiol's role in cognitive processing. These animal studies indicate estradiol impacts cognitive processes supported by regions within dorsolateral prefrontal cortex (DLPFC). Although human functional neuroimaging studies have begun to find similar relationships between estradiol in women for some forms of 'cold' cognitive control, to date no studies have examined the relationship between estradiol and DLPFC function in the context of active attempts to regulate one's emotions. Here, we asked whether peripheral 17-beta estradiol levels in adolescent girls in different pubertal developmental stages (age = 14.9 years ± 1.74) were related to engagement of DLPFC regions during the use of a cognitive strategy for regulating emotion known as reappraisal using functional Magnetic Resonance Imaging. Findings indicated that higher estradiol levels predicted greater DLPFC activity during the down-regulation of negative emotion using reappraisal. This is the first report of an association between estradiol level and DLPFC activity during cognitive reappraisal of negative emotion. The study suggests a possibility that estradiol might positively contribute to regulatory function of a cortical system important for emotional experiences.
Collapse
Affiliation(s)
- Yu Sun Chung
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University.
| | - Andrew Poppe
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University
| | - Stephanie Novotny
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University
| | - C. Neill Epperson
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hedy Kober
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Douglas A. Granger
- Institute for Interdisciplinary Salivary Bioscience Research, University of California at Irvine, Irvine CA; School of Medicine, Bloomberg School of Public Health, and School of Nursing, Johns Hopkins University, Baltimore, MD, USA
| | | | - Kevin Ochsner
- Department of Psychology, Columbia University, New York, NY, USA
| | - James J. Gross
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Godfrey Pearlson
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University.,Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Michael C. Stevens
- Olin Neuropsychiatry Research Center, 200 Retreat Avenue, Whitehall Building- Institute of Living, Hartford, CT, 06106, USA; Department of Psychology and Neuroscience, Duke University.,Department of Psychiatry, Yale University, New Haven, CT, USA
| |
Collapse
|
66
|
Meng ID, Barton ST, Goodney I, Russell R, Mecum NE. Progesterone Application to the Rat Forehead Produces Corneal Antinociception. Invest Ophthalmol Vis Sci 2019; 60:1706-1713. [PMID: 31013343 PMCID: PMC6736375 DOI: 10.1167/iovs.18-26049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Ocular pain and discomfort are the most defining symptoms of dry eye disease. We determined the ability of topical progesterone to affect corneal sensitivity and brainstem processing of nociceptive inputs. Methods Progesterone or vehicle gel was applied to the shaved forehead in male Sprague Dawley rats. As a site control, gel also was applied to the cheek on the side contralateral to corneal stimulation. Corneal mechanical thresholds were determined using the Cochet-Bonnet esthesiometer in intact and lacrimal gland excision–induced dry eye animals. Eye wipe behaviors in response to hypertonic saline and capsaicin were examined, and corneal mustard oil-induced c-Fos immunohistochemistry was quantified in the brainstem spinal trigeminal nucleus. Results Progesterone gel application to the forehead, but not the contralateral cheek, increased corneal mechanical thresholds in intact and lacrimal gland excision animals beginning <30 minutes after treatment. Subcutaneous injection of the local anesthetic bupivacaine into the forehead region before application of progesterone prevented the increase in corneal mechanical thresholds. Furthermore, progesterone decreased capsaicin-evoked eye wipe behavior in intact animals and hypertonic saline evoked eye wipe behavior in dry eye animals. The number of Fos-positive neurons located in the caudal region of the spinal trigeminal nucleus after corneal mustard oil application was reduced in progesterone-treated animals. Conclusions Results from this study indicate that progesterone, when applied to the forehead, produces analgesia as indicated by increased corneal mechanical thresholds and decreased nociceptive responses to hypertonic saline and capsaicin.
Collapse
Affiliation(s)
- Ian D Meng
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States.,Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States
| | - Stephen T Barton
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Ian Goodney
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Rachel Russell
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Neal E Mecum
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States.,Molecular and Biomedical Sciences, University of Maine, Orono, Maine, United States
| |
Collapse
|
67
|
Khadilkar SV, Patil VA. Sex Hormones and Cognition: Where Do We Stand? J Obstet Gynaecol India 2019; 69:303-312. [PMID: 31391735 DOI: 10.1007/s13224-019-01223-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/12/2019] [Indexed: 01/29/2023] Open
Abstract
Hypothalamic-pituitary-gonadal axis regulates the reproductive system. The overall health and wellbeing of a woman is subject to fluctuations in the sex hormones throughout her lifespan. Menopause, either natural or surgically induced, is often associated with cognitive complaints, especially memory disturbances. Sex hormones, besides affecting the reproductive function, affect the central nervous system in many ways. Here, we aim to review the role of sex hormones in cognition and the current evidence on use of or against menopausal hormonal therapy as a cognition enhancer in women with cognitive disturbances, including those with Alzheimer's disease.
Collapse
Affiliation(s)
- Satish V Khadilkar
- Department of Neurology, Bombay Hospital Institute of Medical Sciences, New Marine Lines, Mumbai, Maharashtra 400020 India
| | - Varsha A Patil
- Department of Neurology, Bombay Hospital Institute of Medical Sciences, New Marine Lines, Mumbai, Maharashtra 400020 India
| |
Collapse
|
68
|
Shiono S, Williamson J, Kapur J, Joshi S. Progesterone receptor activation regulates seizure susceptibility. Ann Clin Transl Neurol 2019; 6:1302-1310. [PMID: 31353848 PMCID: PMC6649646 DOI: 10.1002/acn3.50830] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Progesterone is a potent neuromodulator that exerts effects on the brain through neurosteroids, progesterone receptors (PRs), and other molecules. Whether PR activation regulates seizures is not known. We determined whether PR activation increased seizure susceptibility. METHODS Adult female rats that developed epilepsy following lithium-pilocarpine-induced status epilepticus (SE) were used. Seizures were recorded by continuous-video EEG and read by an individual blinded to the treatment of the animals. The animals were treated for a week with progesterone (50 mg/kg per day), and the effect of progesterone withdrawal on seizure frequency was assessed during the subsequent week. During the week of progesterone treatment, the animals were treated with PR antagonist RU-486 (10 mg/kg per day) or a vehicle control, which was administered 30 min before progesterone. In another set of animals, we determined the effect of the PR agonist Nestorone (3 mg/kg per day) on seizure frequency. The animals were treated with Nestorone or vehicle for a week, and seizure frequencies at baseline and during the treatment week were compared. RESULTS Progesterone withdrawal induced twofold increase in seizures in 57% of animals (n = 14). RU-486 treatment in combination with progesterone, prevented this increase, and a smaller fraction of animals (17%) experienced withdrawal seizures (n = 13). The specific activation of PRs by Nestorone also increased the seizure frequency. Forty-six percent (n = 14) of Nestorone-treated animals experienced at least a 50% increase in seizures compared to only 9% of the vehicle-treated animals (n = 11). INTERPRETATION PR activation increased seizure frequency in epileptic animals. Thus, PRs may be novel targets for treating catamenial epilepsy.
Collapse
Affiliation(s)
- Shinnosuke Shiono
- Department of NeurologyUniversity of VirginiaCharlottesvilleVirginia22908
| | - John Williamson
- Department of NeurologyUniversity of VirginiaCharlottesvilleVirginia22908
| | - Jaideep Kapur
- Department of NeurologyUniversity of VirginiaCharlottesvilleVirginia22908
- Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginia22908
- UVA Brain Institute, University of VirginiaCharlottesvilleVirginia22908
| | - Suchitra Joshi
- Department of NeurologyUniversity of VirginiaCharlottesvilleVirginia22908
| |
Collapse
|
69
|
Abdoli A, Rahimi-Bashar F, Torabian S, Sohrabi S, Makarchian HR. Efficacy of Simultaneous Administration of Nimodipine, Progesterone, and Magnesium Sulfate in Patients with Severe Traumatic Brain Injury: A Randomized Controlled Trial. Bull Emerg Trauma 2019; 7:124-129. [PMID: 31198800 PMCID: PMC6555213 DOI: 10.29252/beat-070206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/26/2019] [Accepted: 02/07/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To investigate the safety and efficacy of simultaneous administration of nimodipine, progesterone, magnesium sulfate in patients suffering from severe traumatic brain injury (TBI). METHODS Overall, 90 patients with blunt head trauma who were admitted to the Besat hospital, Hamadan University of Medical Sciences, Iran through the Emergency Department in 2017 to 2018 were randomly assigned to the study or control groups each containing 45 patients. In the study group, intravenous nimodipine 60 mg every 12 hours for 5 days, intramuscular progesterone 1 mg/kg daily for 5 days, and magnesium sulfate 5 grams stat followed by 2.5 grams every 4 hours for 21 days were administered. Daily GCS and jugular venous oxygen saturation (SjvO2) of the patients were measured on admission day (day 0) through hospitalization day 4 at the intensive care unit. Then, all patients were visited at three months after discharge. RESULTS The mean age of the patients was 31.4 ± 12.8 years including 59 (65.6%) men with no significant difference between the groups. The baseline GCS and SjvO2 of the patients were comparable in both groups, however, GCS of the patients in the study group were significantly higher in the next 4 hospitalization days compared to the controls. Whereas, the SjvO2 of the patients were not significantly different between the groups during these days. Three-month mortality rate of the patients in the study group was significantly lower than the three-month mortality rate of the patients in the control groups (22.2% vs. 42.2%, p=0.042). CONCLUSION Administration of combined protocol of magnesium sulfide, progesterone and nimodipine may be safe and effective in patients suffering from severe TBI. CLINICAL TRIAL REGISTRY IRCT201210229534N2.
Collapse
Affiliation(s)
- Ali Abdoli
- Department of Neurosurgery, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Farshid Rahimi-Bashar
- Department of Anesthesiology and Critical Care, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saadat Torabian
- Department of Social Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sepideh Sohrabi
- School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Reza Makarchian
- Department of General Surgery, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
70
|
Tajalli-Nezhad S, Karimian M, Beyer C, Atlasi MA, Azami Tameh A. The regulatory role of Toll-like receptors after ischemic stroke: neurosteroids as TLR modulators with the focus on TLR2/4. Cell Mol Life Sci 2019; 76:523-537. [PMID: 30377701 PMCID: PMC11105485 DOI: 10.1007/s00018-018-2953-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 10/19/2018] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is the most common cerebrovascular disease and considered as a worldwide leading cause of death. After cerebral ischemia, different pathophysiological processes including neuroinflammation, invasion and aggregation of inflammatory cells and up-regulation of cytokines occur simultaneously. In this respect, Toll-like receptors (TLRs) are the first identified important mediators for the activation of the innate immune system and are widely expressed in glial cells and neurons following brain trauma. TLRs are also able to interact with endogenous and exogenous molecules released during ischemia and can increase tissue damage. Particularly, TLR2 and TLR4 activate different downstream inflammatory signaling pathways. In addition, TLR signaling can alternatively play a role for endogenous neuroprotection. In this review, the gene and protein structures, common genetic polymorphisms of TLR2 and TLR4, TLR-related molecular pathways and their putative role after ischemic stroke are delineated. Furthermore, the relationship between neurosteroids and TLRs as neuroprotective mechanism is highlighted in the context of brain ischemia.
Collapse
Affiliation(s)
- Saeedeh Tajalli-Nezhad
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Karimian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
71
|
Joshi S, Kapur J. Neurosteroid regulation of GABA A receptors: A role in catamenial epilepsy. Brain Res 2019; 1703:31-40. [PMID: 29481795 PMCID: PMC6107446 DOI: 10.1016/j.brainres.2018.02.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/08/2017] [Accepted: 02/20/2018] [Indexed: 12/31/2022]
Abstract
The female reproductive hormones progesterone and estrogen regulate network excitability. Fluctuations in the circulating levels of these hormones during the menstrual cycle cause frequent seizures during certain phases of the cycle in women with epilepsy. This seizure exacerbation, called catamenial epilepsy, is a dominant form of drug-refractory epilepsy in women of reproductive age. Progesterone, through its neurosteroid derivative allopregnanolone, increases γ-aminobutyric acid type-A receptor (GABAR)-mediated inhibition in the brain and keeps seizures under control. Catamenial seizures are believed to be a neurosteroid withdrawal symptom, and it was hypothesized that exogenous administration of progesterone to maintain its levels high during luteal phase will treat catamenial seizures. However, in a multicenter, double-blind, phase III clinical trial, progesterone treatment did not suppress catamenial seizures. The expression of GABARs with reduced neurosteroid sensitivity in epileptic animals may explain the failure of the progesterone clinical trial. The expression of neurosteroid-sensitive δ subunit-containing GABARs is reduced, and the expression of α4γ2 subunit-containing GABARs is upregulated, which alters the inhibition of dentate granule cells in epilepsy. These changes reduce the endogenous neurosteroid control of seizures and contribute to catamenial seizures.
Collapse
Affiliation(s)
- Suchitra Joshi
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States.
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, United States
| |
Collapse
|
72
|
Sex differences and the neurobiology of affective disorders. Neuropsychopharmacology 2019; 44:111-128. [PMID: 30061743 PMCID: PMC6235863 DOI: 10.1038/s41386-018-0148-z] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/14/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Abstract
Observations of the disproportionate incidence of depression in women compared with men have long preceded the recent explosion of interest in sex differences. Nonetheless, the source and implications of this epidemiologic sex difference remain unclear, as does the practical significance of the multitude of sex differences that have been reported in brain structure and function. In this article, we attempt to provide a framework for thinking about how sex and reproductive hormones (particularly estradiol as an example) might contribute to affective illness. After briefly reviewing some observed sex differences in depression, we discuss how sex might alter brain function through hormonal effects (both organizational (programmed) and activational (acute)), sex chromosome effects, and the interaction of sex with the environment. We next review sex differences in the brain at the structural, cellular, and network levels. We then focus on how sex and reproductive hormones regulate systems implicated in the pathophysiology of depression, including neuroplasticity, genetic and neural networks, the stress axis, and immune function. Finally, we suggest several models that might explain a sex-dependent differential regulation of affect and susceptibility to affective illness. As a disclaimer, the studies cited in this review are not intended to be comprehensive but rather serve as examples of the multitude of levels at which sex and reproductive hormones regulate brain structure and function. As such and despite our current ignorance regarding both the ontogeny of affective illness and the impact of sex on that ontogeny, sex differences may provide a lens through which we may better view the mechanisms underlying affective regulation and dysfunction.
Collapse
|
73
|
Thaung Zaw JJ, Howe PRC, Wong RHX. Postmenopausal health interventions: Time to move on from the Women's Health Initiative? Ageing Res Rev 2018; 48:79-86. [PMID: 30355506 DOI: 10.1016/j.arr.2018.10.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/25/2018] [Accepted: 10/17/2018] [Indexed: 11/15/2022]
Abstract
Menopause is a critical period during which, without timely interventions, increased risks of cardiovascular and metabolic diseases, osteoporosis, sexual dysfunction and premature cognitive decline will contribute to diminished quality-of-life in women. Hormone therapy (HT) used to be the standard of care for managing vasomotor symptoms and prevention of chronic diseases until publication of the Women's Health Initiative (WHI) in 2002. Concerned about risks highlighted in WHI publications, many symptomatic women promptly ceased HT which resulted in increased vasomotor symptoms, osteoporosis-related-fractures and insomnia. Data from post-hoc WHI analyses and newer clinical trials consistently show reductions in coronary heart disease and mortality when estrogen therapy is initiated soon after menopause, whereas administration in later years and/or in combination with progesterone carries increased risks. However, no validated primary preventive strategies are available for younger postmenopausal women (<60 years), highlighting the need to re-evaluate the use of estrogen alone for which the risk-benefit balance appears positive. In contrast, in older women (>60 years), risks associated with oral HT exceed benefits; however transdermal estrogen may offer a safer alternative and should be further evaluated. Alternative therapies such as phytoestrogens and non-hormonal prescriptions may be beneficial for older women or those who are unsuitable for HT. Long-term head-to-head comparisons of HT with alternative interventions are warranted to confirm their efficacy for chronic disease prevention.
Collapse
Affiliation(s)
- Jay Jay Thaung Zaw
- Clinical Nutrition Research Centre, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, 2308, Australia
| | - Peter Ranald Charles Howe
- Clinical Nutrition Research Centre, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, 2308, Australia; University of Southern Queensland, Institute for Resilient Regions, Springfield Central, Queensland, 4300, Australia
| | - Rachel Heloise Xiwen Wong
- Clinical Nutrition Research Centre, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, 2308, Australia; University of Southern Queensland, Institute for Resilient Regions, Springfield Central, Queensland, 4300, Australia.
| |
Collapse
|
74
|
El Amki M, Binder N, Steffen R, Schneider H, Luft AR, Weller M, Imthurn B, Merki-Feld GS, Wegener S. Contraceptive drugs mitigate experimental stroke-induced brain injury. Cardiovasc Res 2018; 115:637-646. [DOI: 10.1093/cvr/cvy248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
AbstractAimsEffective stroke treatments beyond reperfusion remain scant. The natural steroid hormone progesterone has shown protective effects in experimental models of brain injury and cardiovascular disease. However, unfavourable bioavailability limits its clinical use. Desogestrel and drospirenone are new generation progestins with progesterone-like properties, developed as oral contraceptives with excellent bioavailability and safety profile. We investigated the neuroprotective properties of these progestins in vivo using transient middle cerebral artery occlusion (MCAO) and in vitro using an oxygen-glucose deprivation and reoxygenation (OGD/R) model in primary neuronal cells.Methods and resultsMCAO was induced in female, female ovariectomized (modelling postmenopausal females) and male mice. Treatment with the progestins resulted in less severe strokes after MCAO and less neuronal death in OGD/R. Desogestrel and drospirenone induced higher expression levels of GABAAR α4 and delta subunits within the brain, suggesting changes in GABAAR configuration favouring tonic inhibition as potential mechanism of action. Treatment with the GABAAR blocker picrotoxin abolished the protection afforded by the progestins in vivo and in vitro.ConclusionFor the first time, here, we delineate a potential role of desogestrel and drospirenone, both clinically approved and safe drugs in mitigating the consequences of stroke. Contraception with desogestrel and drospirenone in progestin-only preparations may be particularly beneficial for women at risk of stroke.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Nadine Binder
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Riccardo Steffen
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Hannah Schneider
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Andreas R Luft
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Bruno Imthurn
- Department of Reproductive Endocrinology, University Hospital Zurich, Switzerland
| | | | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| |
Collapse
|
75
|
Ghoreishi Z, Keshavarz S, Asghari Jafarabadi M, Fathifar Z, Goodman KA, Esfahani A. Risk factors for paclitaxel-induced peripheral neuropathy in patients with breast cancer. BMC Cancer 2018; 18:958. [PMID: 30290775 PMCID: PMC6173931 DOI: 10.1186/s12885-018-4869-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/26/2018] [Indexed: 11/10/2022] Open
Abstract
Background Paclitaxel induced peripheral neuropathy (PIPN) is a major debilitating side effect of paclitaxel in patients with breast cancer with no fully known mechanisms. The aim of the study was to find out the possible risk factors for PIPN. Methods Eligible patients with node positive breast cancer undergoing chemotherapy with paclitaxel were assessed. They belonged to an initial randomized controlled trial in which the effectiveness of omega-3 fatty acids in preventing and reducing severity of PIPN was evaluated (protocol ID: NCT01049295). Reduced total neuropathy score (r-TNS) was used for measuring PIPN. All analyses were performed adjusting for intervention effect. The association between age, BMI, BSA, pathological grade, molecular biomarkers and PIPN was evaluated. Results Fifty-seven patients with breast cancer were investigated. Age was significantly associated with risk of PIPN (RR:1.50, P value = .024). Body mass index and BSA had significant association with severity of PIPN (B:1.28, P = .025; and B: 3.88, P = .010 respectively). Also, BSA showed a significant association with the risk of PIPN (RR: 2.28, P = .035; B: 3.88, P = .035). Incidence and severity of PIPN were much more pronounced in progesterone receptor positive (PR+) patients (RR:1.88, P = .015 and B:1.54, P = .012). Multivariate analysis showed that age and the status of PR+ were independent risk factor for incidence and the status of PR+ was the only independent risk factor for severity of PIPN. Conclusion Age, BSA and the status of PR+, should be considered as the risk factors for PIPN before commencement of chemotherapy with paclitaxel in patients with breast cancer. Older patients, those with greater BSA and PR+ patients may need closer follow up and more medical attention due to greater incidence and severity of PIPN.
Collapse
Affiliation(s)
- Zohreh Ghoreishi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedali Keshavarz
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zahra Fathifar
- School of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karyn A Goodman
- Department of Radiation Oncology, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Ali Esfahani
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Shahid Ghazi Hospital, Tabriz, Iran.
| |
Collapse
|
76
|
Gurvich C, Gavrilidis E, Worsley R, Hudaib A, Thomas N, Kulkarni J. Menstrual cycle irregularity and menopause status influence cognition in women with schizophrenia. Psychoneuroendocrinology 2018; 96:173-178. [PMID: 29980009 DOI: 10.1016/j.psyneuen.2018.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/12/2018] [Accepted: 06/27/2018] [Indexed: 01/19/2023]
Abstract
Cognitive impairments are a core feature of schizophrenia and contribute significantly to functional complications. Current pharmacological treatments do not ameliorate cognitive dysfunction and the aetiology of cognitive impairments are poorly understood. Hormones of the hypothalamic-pituitary-gonadal (HPG) axis that regulate reproductive function have multiple effects on the development, maintenance and function of the brain and have been suggested to also influence cognition. The aim of the current study was to investigate how HPG axis hormones effect cognition, specifically exploring the influence of menopause status and menstrual cycle irregularity on cognitive performance in women with schizophrenia. The data for the present study represents pooled baseline data from three clinical trials. Two hundred and forty female participants with a diagnosis of schizophrenia or schizoaffective disorder were included in the analysis. Cognition was assessed using the Repeatable Battery for the Assessment of Neuropsychological Status. Hormone assays for serum sex steroids and pituitary hormones (including estradiol, progesterone, luteinising hormone and follicle-stimulating hormone) were conducted and women were classified as postmenopausal; perimenopausal; premenopausal/reproductive, further classified into regular and irregular menstrual cycles. To model a comparison of cognitive performance for i) perimenopausal; ii) post-menopausal women and iii) reproductive aged women with irregular cycles to reproductive aged women with regular cycles a semiparametric regression model (generalised additive mode) was fitted. The results revealed that in females with schizophrenia, menstrual cycle irregularity predicted significantly poorer cognitive performance in the areas of psychomotor speed, verbal fluency and verbal memory. Perimenopause was not associated with cognitive changes and the post-menopausal period was associated with poorer visuospatial performance. This study provides evidence to associate reproductive hormones with cognitive dysfunction in schizophrenia.
Collapse
Affiliation(s)
- C Gurvich
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Melbourne, Australia.
| | - E Gavrilidis
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Melbourne, Australia
| | - R Worsley
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Melbourne, Australia
| | - A Hudaib
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Melbourne, Australia
| | - N Thomas
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Melbourne, Australia
| | - J Kulkarni
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Melbourne, Australia
| |
Collapse
|
77
|
Gurvich C, Hoy K, Thomas N, Kulkarni J. Sex Differences and the Influence of Sex Hormones on Cognition through Adulthood and the Aging Process. Brain Sci 2018; 8:brainsci8090163. [PMID: 30154388 PMCID: PMC6162653 DOI: 10.3390/brainsci8090163] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 12/28/2022] Open
Abstract
Hormones of the hypothalamic-pituitary-gonadal (HPG) axis that regulate reproductive function have multiple effects on the development, maintenance and function of the brain. Sex differences in cognitive functioning have been reported in both health and disease, which may be partly attributed to sex hormones. The aim of the current paper was to provide a theoretical review of how sex hormones influence cognitive functioning across the lifespan as well as provide an overview of the literature on sex differences and the role of sex hormones in cognitive decline, specifically in relation to Alzheimer’s disease (AD). A summary of current hormone and sex-based interventions for enhancing cognitive functioning and/or reducing the risk of Alzheimer’s disease is also provided.
Collapse
Affiliation(s)
- Caroline Gurvich
- Monash Alfred Psychiatry Research Centre, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC 3004, Australia.
| | - Kate Hoy
- Monash Alfred Psychiatry Research Centre, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC 3004, Australia.
| | - Natalie Thomas
- Monash Alfred Psychiatry Research Centre, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC 3004, Australia.
| | - Jayashri Kulkarni
- Monash Alfred Psychiatry Research Centre, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC 3004, Australia.
| |
Collapse
|
78
|
Vargas DM, De Bastiani MA, Zimmer ER, Klamt F. Alzheimer's disease master regulators analysis: search for potential molecular targets and drug repositioning candidates. ALZHEIMERS RESEARCH & THERAPY 2018; 10:59. [PMID: 29935546 PMCID: PMC6015462 DOI: 10.1186/s13195-018-0394-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/30/2018] [Indexed: 02/03/2023]
Abstract
Background Alzheimer’s disease (AD) is a multifactorial and complex neuropathology that involves impairment of many intricate molecular mechanisms. Despite recent advances, AD pathophysiological characterization remains incomplete, which hampers the development of effective treatments. In fact, currently, there are no effective pharmacological treatments for AD. Integrative strategies such as transcription regulatory network and master regulator analyses exemplify promising new approaches to study complex diseases and may help in the identification of potential pharmacological targets. Methods In this study, we used transcription regulatory network and master regulator analyses on transcriptomic data of human hippocampus to identify transcription factors (TFs) that can potentially act as master regulators in AD. All expression profiles were obtained from the Gene Expression Omnibus database using the GEOquery package. A normal hippocampus transcription factor-centered regulatory network was reconstructed using the ARACNe algorithm. Master regulator analysis and two-tail gene set enrichment analysis were employed to evaluate the inferred regulatory units in AD case-control studies. Finally, we used a connectivity map adaptation to prospect new potential therapeutic interventions by drug repurposing. Results We identified TFs with already reported involvement in AD, such as ATF2 and PARK2, as well as possible new targets for future investigations, such as CNOT7, CSRNP2, SLC30A9, and TSC22D1. Furthermore, Connectivity Map Analysis adaptation suggested the repositioning of six FDA-approved drugs that can potentially modulate master regulator candidate regulatory units (Cefuroxime, Cyproterone, Dydrogesterone, Metrizamide, Trimethadione, and Vorinostat). Conclusions Using a transcription factor-centered regulatory network reconstruction we were able to identify several potential molecular targets and six drug candidates for repositioning in AD. Our study provides further support for the use of bioinformatics tools as exploratory strategies in neurodegenerative diseases research, and also provides new perspectives on molecular targets and drug therapies for future investigation and validation in AD. Electronic supplementary material The online version of this article (10.1186/s13195-018-0394-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- D M Vargas
- Laboratory of Cellular Biochemistry, Biochemistry Department, Institute of Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil.
| | - M A De Bastiani
- Laboratory of Cellular Biochemistry, Biochemistry Department, Institute of Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - E R Zimmer
- Pharmacology Department, Institute of Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil.,Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, 90619-900, Brazil
| | - F Klamt
- Laboratory of Cellular Biochemistry, Biochemistry Department, Institute of Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil.,National Science Technology Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq), Porto Alegre, Brazil
| |
Collapse
|
79
|
Allitt BJ, Johnstone VPA, Richards KL, Yan EB, Rajan R. Progesterone Sharpens Temporal Response Profiles of Sensory Cortical Neurons in Animals Exposed to Traumatic Brain Injury. Cell Transplant 2018; 26:1202-1223. [PMID: 28933224 PMCID: PMC5657734 DOI: 10.1177/0963689717714326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) initiates a cascade of pathophysiological changes that are both complex and difficult to treat. Progesterone (P4) is a neuroprotective treatment option that has shown excellent preclinical benefits in the treatment of TBI, but these benefits have not translated well in the clinic. We have previously shown that P4 exacerbates the already hypoactive upper cortical responses in the short-term post-TBI and does not reduce upper cortical hyperactivity in the long term, and we concluded that there is no tangible benefit to sensory cortex firing strength. Here we examined the effects of P4 treatment on temporal coding resolution in the rodent sensory cortex in both the short term (4 d) and long term (8 wk) following impact-acceleration–induced TBI. We show that in the short-term postinjury, TBI has no effect on sensory cortex temporal resolution and that P4 also sharpens the response profile in all cortical layers in the uninjured brain and all layers other than layer 2 (L2) in the injured brain. In the long term, TBI broadens the response profile in all cortical layers despite firing rate hyperactivity being localized to upper cortical layers and P4 sharpens the response profile in TBI animals in all layers other than L2 and has no long-term effect in the sham brain. These results indicate that P4 has long-term effects on sensory coding that may translate to beneficial perceptual outcomes. The effects seen here, combined with previous beneficial preclinical data, emphasize that P4 is still a potential treatment option in ameliorating TBI-induced disorders.
Collapse
Affiliation(s)
- Benjamin J Allitt
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Victoria P A Johnstone
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia.,2 School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia, Australia
| | - Katrina L Richards
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Edwin B Yan
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Ramesh Rajan
- 1 Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
80
|
Yao Q, Feng M, Yang B, Long Z, Luo S, Luo M, He G, Wang K. Effects of ovarian hormone loss on neuritic plaques and autophagic flux in the brains of adult female APP/PS1 double-transgenic mice. Acta Biochim Biophys Sin (Shanghai) 2018; 50:447-455. [PMID: 29617703 PMCID: PMC5946928 DOI: 10.1093/abbs/gmy032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/07/2018] [Indexed: 01/07/2023] Open
Abstract
Epidemiologic studies have demonstrated that women account for two-thirds of Alzheimer’s disease (AD) cases, for which the decline in circulating gonadal hormone is considered to be one of the major risk factors. In addition, ovarian hormone deficiency may affect β-amyloid (Aβ) deposition, which has a close relationship with autophagic flux. In this study, we investigated the impact of short-term or long-term ovarian hormone deprivation on two mouse models, the non-transgenic (wild-type) and the APP/PS1 double-transgenic AD (2×TgAD) model. Autophagy-related proteins (Beclin1, LC3, and p62) and lysosome-related proteins were detected to evaluate Aβ deposition and autophagy. Our results showed that in the group with short-term depletion of ovarian hormones by ovariectomy (ovx), Beclin1, Cathepsin B (Cath-B), and LAMP1 levels were significantly decreased, while the levels of LC3-II and p62 were increased. In the long-term group, however, there was a sharp decline in Beclin1, LC3-II, Cath-B, and LAMP1 expression but not in p62 expression which is increased. It is worthwhile to note that the occurrence of neuritic plaque-induced ovarian hormone loss increased both the Aβ level and neuritic plaque deposition in 2×TgAD mice. Therefore, autophagy may play an important role in the pathogenesis of female AD, which is also expected to help post-menopausal patients with AD.
Collapse
Affiliation(s)
- Qiuhui Yao
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Min Feng
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Bo Yang
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Zhimin Long
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Shifang Luo
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Min Luo
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Guiqiong He
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Kejian Wang
- Chongqing Key Laboratory of Neurobiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
- Correspondence address. Tel: +86-23-68485763; Fax: +86-23-68485000; E-mail:
| |
Collapse
|
81
|
Joshi S, Sun H, Rajasekaran K, Williamson J, Perez-Reyes E, Kapur J. A novel therapeutic approach for treatment of catamenial epilepsy. Neurobiol Dis 2018; 111:127-137. [PMID: 29274741 PMCID: PMC5803337 DOI: 10.1016/j.nbd.2017.12.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/27/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022] Open
Abstract
Many women with epilepsy experience perimenstrual seizure exacerbation, referred to as catamenial epilepsy. There is no effective treatment for this condition, proposed to result from withdrawal of neurosteroid-mediated effects of progesterone. A double-blind, multicenter, phase III, clinical trial of catamenial epilepsy has failed to find a beneficial effect of progesterone. The neurosteroid-mediated effects of progesterone have been extensively studied in relation to catamenial epilepsy; however, the effects mediated by progesterone receptor activation have been overlooked. We determined whether progesterone increased excitatory transmission in the hippocampus via activation of progesterone receptors, which may play a role in regulating catamenial seizure exacerbation. In a double-blind study using a rat model of catamenial epilepsy, we found that treatment with RU-486, which blocks progesterone and glucocorticoid receptors, significantly attenuated neurosteroid withdrawal-induced seizures. Furthermore, progesterone treatment as well as endogenous rise in progesterone during estrous cycle increased the expression of GluA1 and GluA2 subunits of AMPA receptors in the hippocampi, and enhanced the AMPA receptor-mediated synaptic transmission of CA1 pyramidal neurons. The progesterone-induced plasticity of AMPA receptors was blocked by RU-486 treatment and progesterone also failed to increase AMPA receptor expression in progesterone receptor knockout mice. These studies demonstrate that progesterone receptor activation regulates AMPA receptor expression and may play a role in catamenial seizure exacerbation.
Collapse
Affiliation(s)
- Suchitra Joshi
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - Huayu Sun
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - Karthik Rajasekaran
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - John Williamson
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, United States
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, United States.
| |
Collapse
|
82
|
Adhya D, Annuario E, Lancaster MA, Price J, Baron‐Cohen S, Srivastava DP. Understanding the role of steroids in typical and atypical brain development: Advantages of using a "brain in a dish" approach. J Neuroendocrinol 2018; 30:e12547. [PMID: 29024164 PMCID: PMC5838783 DOI: 10.1111/jne.12547] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/14/2017] [Accepted: 10/03/2017] [Indexed: 01/02/2023]
Abstract
Steroids have an important role in growth, development, sexual differentiation and reproduction. All four classes of steroids, androgens, oestrogens, progestogens and glucocorticoids, have varying effects on the brain. Androgens and oestrogens are involved in the sexual differentiation of the brain, and also influence cognition. Progestogens such as progesterone and its metabolites have been shown to be involved in neuroprotection, although their protective effects are timing-dependent. Glucocorticoids are linked with stress and memory performance, also in a dose- and time-dependent manner. Importantly, dysfunction in steroid function has been implicated in the pathogenesis of disease. Moreover, regulating steroid-signalling has been suggested as potential therapeutic avenue for the treatment of a number of neurodevelopmental, psychiatric and neurodegenerative disorders. Therefore, clarifying the role of steroids in typical and atypical brain function is essential for understanding typical brain functions, as well as determining their potential use for pharmacological intervention in the atypical brain. However, the majority of studies have thus far have been conducted using animal models, with limited work using native human tissue or cells. Here, we review the effect of steroids in the typical and atypical brain, focusing on the cellular, molecular functions of these molecules determined from animal models, and the therapeutic potential as highlighted by human studies. We further discuss the promise of human-induced pluripotent stem cells, including advantages of using three-dimensional neuronal cultures (organoids) in high-throughput screens, in accelerating our understanding of the role of steroids in the typical brain, and also with respect to their therapeutic value in the understanding and treatment of the atypical brain.
Collapse
Affiliation(s)
- D. Adhya
- Department of PsychiatryAutism Research CentreUniversity of CambridgeCambridgeUK
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Laboratory of Molecular BiologyCambridgeUK
| | - E. Annuario
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | | | - J. Price
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
- National Institute for Biological Standards and ControlSouth MimmsUK
| | - S. Baron‐Cohen
- Department of PsychiatryAutism Research CentreUniversity of CambridgeCambridgeUK
| | - D. P. Srivastava
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| |
Collapse
|
83
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2018. [PMID: 29311911 DOI: 10.3389/fnagi.2017.00430/xml/nlm] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
84
|
Nuzzi R, Scalabrin S, Becco A, Panzica G. Gonadal Hormones and Retinal Disorders: A Review. Front Endocrinol (Lausanne) 2018; 9:66. [PMID: 29551993 PMCID: PMC5840201 DOI: 10.3389/fendo.2018.00066] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/14/2018] [Indexed: 12/27/2022] Open
Abstract
AIM Gonadal hormones are essential for reproductive function, but can act on neural and other organ systems, and are probably the cause of the large majority of known sex differences in function and disease. The aim of this review is to provide evidence for this hypothesis in relation to eye disorders and to retinopathies in particular. METHODS Epidemiological studies and research articles were reviewed. RESULTS Analysis of the biological basis for a relationship between eye diseases and hormones showed that estrogen, androgen, and progesterone receptors are present throughout the eye and that these steroids are locally produced in ocular tissues. Sex hormones can have a neuroprotective action on the retina and modulate ocular blood flow. There are differences between the male and the female retina; moreover, sex hormones can influence the development (or not) of certain disorders. For example, exposure to endogenous estrogens, depending on age at menarche and menopause and number of pregnancies, and exposure to exogenous estrogens, as in hormone replacement therapy and use of oral contraceptives, appear to protect against age-related macular degeneration (both drusenoid and neurovascular types), whereas exogenous testosterone therapy is a risk factor for central serous chorioretinopathy. Macular hole is more common among women than men, particularly in postmenopausal women probably owing to the sudden drop in estrogen production in later middle age. Progestin therapy appears to ameliorate the course of retinitis pigmentosa. Diabetic retinopathy, a complication of diabetes, may be more common among men than women. CONCLUSION We observed a correlation between many retinopathies and sex, probably as a result of the protective effect some gonadal hormones may exert against the development of certain disorders. This may have ramifications for the use of hormone therapy in the treatment of eye disease and of retinal disorders in particular.
Collapse
Affiliation(s)
- Raffaele Nuzzi
- Eye Clinic, Department of Surgical Sciences, University of Turin, Turin, Italy
- *Correspondence: Raffaele Nuzzi,
| | - Simona Scalabrin
- Eye Clinic, Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Alice Becco
- Eye Clinic, Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Giancarlo Panzica
- Laboratory of Neuroendocrinology, Department of Neuroscience Rita Levi-Montalcini, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri-Ottolenghi (NICO), Orbassano, Italy
| |
Collapse
|
85
|
Muñoz-Mayorga D, Guerra-Araiza C, Torner L, Morales T. Tau Phosphorylation in Female Neurodegeneration: Role of Estrogens, Progesterone, and Prolactin. Front Endocrinol (Lausanne) 2018; 9:133. [PMID: 29643836 PMCID: PMC5882780 DOI: 10.3389/fendo.2018.00133] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
Sex differences are important to consider when studying different psychiatric, neurodevelopmental, and neurodegenerative disorders, including Alzheimer's disease (AD). These disorders can be affected by dimorphic changes in the central nervous system and be influenced by sex-specific hormones and neuroactive steroids. In fact, AD is more prevalent in women than in men. One of the main characteristics of AD is the formation of neurofibrillary tangles, composed of the phosphoprotein Tau, and neuronal loss in specific brain regions. The scope of this work is to review the existing evidence on how a set of hormones (estrogen, progesterone, and prolactin) affect tau phosphorylation in the brain of females under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Daniel Muñoz-Mayorga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
- *Correspondence: Teresa Morales,
| |
Collapse
|
86
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2017; 9:430. [PMID: 29311911 PMCID: PMC5743731 DOI: 10.3389/fnagi.2017.00430] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
87
|
Murray SR, Stock SJ, Norman JE. Long-term childhood outcomes after interventions for prevention and management of preterm birth. Semin Perinatol 2017; 41:519-527. [PMID: 29191292 DOI: 10.1053/j.semperi.2017.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Globally, preterm birth rates are rising and have a significant impact on neonatal morbidity and mortality. Preterm birth remains difficult to prevent and a number of strategies for preterm birth prevention (progesterone, cervical pessaries, cervical cerclage, tocolytics, and antibiotics) have been identified. While some of these show more promise, there is a paucity of evidence regarding the long-term effects of these strategies on childhood outcomes. Strategies used to improve the health of babies if born preterm, such as antenatal magnesium sulfate for fetal neuroprotection and antenatal corticosteroids for fetal lung maturation, show evidence of short-term benefit but lack large-scale follow-up data of long-term childhood outcomes. Future research on preterm birth interventions should include long-term follow-up of the children, ideally with similar outcome measures to allow for future meta-analyses.
Collapse
Affiliation(s)
- Sarah R Murray
- Tommy's Centre for Maternal and Fetal Health, MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Sarah J Stock
- Tommy's Centre for Maternal and Fetal Health, MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jane E Norman
- Tommy's Centre for Maternal and Fetal Health, MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
88
|
Nikolić T, Petronijević M, Sopta J, Velimirović M, Stojković T, Jevtić Dožudić G, Aksić M, Radonjić NV, Petronijević N. Haloperidol affects bones while clozapine alters metabolic parameters - sex specific effects in rats perinatally treated with phencyclidine. BMC Pharmacol Toxicol 2017; 18:65. [PMID: 29020988 PMCID: PMC5637335 DOI: 10.1186/s40360-017-0171-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 10/03/2017] [Indexed: 01/26/2023] Open
Abstract
Background The presentation of schizophrenia (SCH) symptoms differs between the sexes. Long-term treatment with antipsychotics is frequently associated with decreased bone mineral density, increased fracture risk and metabolic side effects. Perinatal phencyclidine (PCP) administration to rodents represents an animal model of SCH. The aim of this study was to assess the effects of chronic haloperidol and clozapine treatment on bone mass, body composition, corticosterone, IL-6 and TNF-α concentrations and metabolic parameters in male and female rats perinatally treated with PCP. Methods Six groups of male and six groups of female rats (n = 6-12 per group) were subcutaneously treated on 2nd, 6th, 9th and 12th postnatal day (PN), with either PCP (10 mg/kg) or saline. At PN35, one NaCl and PCP group (NaCl-H and PCP-H) started receiving haloperidol (1 mg/kg/day) and one NaCl and PCP group (NaCl-C and PCP-C) started receiving clozapine (20 mg/kg/day) dissolved in drinking water. The remaining NaCl and PCP groups received water. Dual X-ray absorptiometry measurements were performed on PN60 and PN98. Animals were sacrificed on PN100. Femur was analysed by light microscopy. Concentrations of corticosterone, TNF-α and IL-6 were measured in serum samples using enzyme-linked immunosorbent assay (ELISA) commercially available kits. Glucose, cholesterol and triacylglycerol concentrations were measured in serum spectrophotometrically. Results Our results showed that perinatal PCP administration causes a significant decrease in bone mass and deterioration in bone quality in male and female rats. Haloperidol had deleterious, while clozapine had protective effect on bones. The effects of haloperidol on bones were more pronounced in male rats. It seems that the observed changes are not the consequence of the alterations of corticosterone, IL-6 and TNF-α concentration since no change of these factors was observed. Clozapine induced increase of body weight and retroperitoneal fat in male rats regardless of perinatal treatment. Furthermore, clozapine treatment caused sex specific increase in pro-inflammatory cytokines. Conclusion Taken together our findings confirm that antipsychotics have complex influence on bone and metabolism. Evaluation of potential markers for individual risk of antipsychotics induced adverse effects could be valuable for improvement of therapy of this life-long lasting disease. Electronic supplementary material The online version of this article (10.1186/s40360-017-0171-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tatjana Nikolić
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Petronijević
- Military Medical Academy, Clinic of Rheumatology, University of Defence, Belgrade, Serbia
| | - Jelena Sopta
- Institute of Pathology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milica Velimirović
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tihomir Stojković
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Gordana Jevtić Dožudić
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Aksić
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nevena V Radonjić
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Nataša Petronijević
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
89
|
Engler-Chiurazzi EB, Brown CM, Povroznik JM, Simpkins JW. Estrogens as neuroprotectants: Estrogenic actions in the context of cognitive aging and brain injury. Prog Neurobiol 2017; 157:188-211. [PMID: 26891883 PMCID: PMC4985492 DOI: 10.1016/j.pneurobio.2015.12.008] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/06/2015] [Accepted: 12/10/2015] [Indexed: 12/30/2022]
Abstract
There is ample empirical evidence to support the notion that the biological impacts of estrogen extend beyond the gonads to other bodily systems, including the brain and behavior. Converging preclinical findings have indicated a neuroprotective role for estrogen in a variety of experimental models of cognitive function and brain insult. However, the surprising null or even detrimental findings of several large clinical trials evaluating the ability of estrogen-containing hormone treatments to protect against age-related brain changes and insults, including cognitive aging and brain injury, led to hesitation by both clinicians and patients in the use of exogenous estrogenic treatments for nervous system outcomes. That estrogen-containing therapies are used by tens of millions of women for a variety of health-related applications across the lifespan has made identifying conditions under which benefits with estrogen treatment will be realized an important public health issue. Here we provide a summary of the biological actions of estrogen and estrogen-containing formulations in the context of aging, cognition, stroke, and traumatic brain injury. We have devoted special attention to highlighting the notion that estrogen appears to be a conditional neuroprotectant whose efficacy is modulated by several interacting factors. By developing criteria standards for desired beneficial peripheral and neuroprotective outcomes among unique patient populations, we can optimize estrogen treatments for attenuating the consequences of, and perhaps even preventing, cognitive aging and brain injury.
Collapse
Affiliation(s)
- E B Engler-Chiurazzi
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| | - C M Brown
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Neurobiology and Anatomy, West Virginia University, Morgantown, WV 26506, United States.
| | - J M Povroznik
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Pediatrics, West Virginia University, Morgantown, WV 26506, United States.
| | - J W Simpkins
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| |
Collapse
|
90
|
Danford ID, Verkuil LD, Choi DJ, Collins DW, Gudiseva HV, Uyhazi KE, Lau MK, Kanu LN, Grant GR, Chavali VRM, O'Brien JM. Characterizing the "POAGome": A bioinformatics-driven approach to primary open-angle glaucoma. Prog Retin Eye Res 2017; 58:89-114. [PMID: 28223208 PMCID: PMC5464971 DOI: 10.1016/j.preteyeres.2017.02.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/03/2017] [Accepted: 02/10/2017] [Indexed: 01/10/2023]
Abstract
Primary open-angle glaucoma (POAG) is a genetically, physiologically, and phenotypically complex neurodegenerative disorder. This study addressed the expanding collection of genes associated with POAG, referred to as the "POAGome." We used bioinformatics tools to perform an extensive, systematic literature search and compiled 542 genes with confirmed associations with POAG and its related phenotypes (normal tension glaucoma, ocular hypertension, juvenile open-angle glaucoma, and primary congenital glaucoma). The genes were classified according to their associated ocular tissues and phenotypes, and functional annotation and pathway analyses were subsequently performed. Our study reveals that no single molecular pathway can encompass the pathophysiology of POAG. The analyses suggested that inflammation and senescence may play pivotal roles in both the development and perpetuation of the retinal ganglion cell degeneration seen in POAG. The TGF-β signaling pathway was repeatedly implicated in our analyses, suggesting that it may be an important contributor to the manifestation of POAG in the anterior and posterior segments of the globe. We propose a molecular model of POAG revolving around TGF-β signaling, which incorporates the roles of inflammation and senescence in this disease. Finally, we highlight emerging molecular therapies that show promise for treating POAG.
Collapse
Affiliation(s)
- Ian D Danford
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lana D Verkuil
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel J Choi
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David W Collins
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Harini V Gudiseva
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katherine E Uyhazi
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Marisa K Lau
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Levi N Kanu
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gregory R Grant
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA, Penn Center for Bioinformatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Venkata R M Chavali
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Joan M O'Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
91
|
Brotfain E, Gruenbaum SE, Boyko M, Kutz R, Zlotnik A, Klein M. Neuroprotection by Estrogen and Progesterone in Traumatic Brain Injury and Spinal Cord Injury. Curr Neuropharmacol 2017; 14:641-53. [PMID: 26955967 PMCID: PMC4981744 DOI: 10.2174/1570159x14666160309123554] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 12/31/2015] [Accepted: 02/25/2016] [Indexed: 12/25/2022] Open
Abstract
In recent years there has been a growing body of clinical and laboratory evidence demonstrating the neuroprotective effects of estrogen and progesterone after traumatic brain injury (TBI) and spinal cord injury (SCI). In humans, women have been shown to have a lower incidence of morbidity and mortality after TBI compared with age-matched men. Similarly, numerous laboratory studies have demonstrated that estrogen and progesterone administration is associated with a mortality reduction, improvement in neurological outcomes, and a reduction in neuronal apoptosis after TBI and SCI. Here, we review the evidence that supports hormone-related neuroprotection and discuss possible underlying mechanisms. Estrogen and progesterone-mediated neuroprotection are thought to be related to their effects on hormone receptors, signaling systems, direct antioxidant effects, effects on astrocytes and microglia, modulation of the inflammatory response, effects on cerebral blood flow and metabolism, and effects on mediating glutamate excitotoxicity. Future laboratory research is needed to better determine the mechanisms underlying the hormones' neuroprotective effects, which will allow for more clinical studies. Furthermore, large randomized clinical control trials are needed to better assess their role in human neurodegenerative conditions.
Collapse
Affiliation(s)
- Evgeni Brotfain
- Department of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | | | | | |
Collapse
|
92
|
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of death and disability, and the identification of effective, inexpensive and widely practicable treatments for brain injury is of great public health importance worldwide. Progesterone is a naturally produced hormone that has well-defined pharmacokinetics, is widely available, inexpensive, and has steroidal, neuroactive and neurosteroidal actions in the central nervous system. It is, therefore, a potential candidate for treating TBI patients. However, uncertainty exists regarding the efficacy of this treatment. This is an update of our previous review of the same title, published in 2012. OBJECTIVES To assess the effects of progesterone on neurologic outcome, mortality and disability in patients with acute TBI. To assess the safety of progesterone in patients with acute TBI. SEARCH METHODS We updated our searches of the following databases: the Cochrane Injuries Group's Specialised Register (30 September 2016), the Cochrane Central Register of Controlled Trials (CENTRAL; Issue 9, 2016), MEDLINE (Ovid; 1950 to 30 September 2016), Embase (Ovid; 1980 to 30 September 2016), Web of Science Core Collection: Conference Proceedings Citation Index-Science (CPCI-S; 1990 to 30 September 2016); and trials registries: Clinicaltrials.gov (30 September 2016) and the World Health Organization (WHO) International Clinical Trials Registry Platform (30 September 2016). SELECTION CRITERIA We included randomised controlled trials (RCTs) of progesterone versus no progesterone (or placebo) for the treatment of people with acute TBI. DATA COLLECTION AND ANALYSIS Two review authors screened search results independently to identify potentially relevant studies for inclusion. Independently, two review authors selected trials that met the inclusion criteria from the results of the screened searches, with no disagreement. MAIN RESULTS We included five RCTs in the review, with a total of 2392 participants. We assessed one trial to be at low risk of bias; two at unclear risk of bias (in one multicentred trial the possibility of centre effects was unclear, whilst the other trial was stopped early), and two at high risk of bias, due to issues with blinding and selective reporting of outcome data.All included studies reported the effects of progesterone on mortality and disability. Low quality evidence revealed no evidence of a difference in overall mortality between the progesterone group and placebo group (RR 0.91, 95% CI 0.65 to 1.28, I² = 62%; 5 studies, 2392 participants, 2376 pooled for analysis). Using the GRADE criteria, we assessed the quality of the evidence as low, due to the substantial inconsistency across studies.There was also no evidence of a difference in disability (unfavourable outcomes as assessed by the Glasgow Outcome Score) between the progesterone group and placebo group (RR 0.98, 95% CI 0.89 to 1.06, I² = 37%; 4 studies; 2336 participants, 2260 pooled for analysis). We assessed the quality of this evidence to be moderate, due to inconsistency across studies.Data were not available for meta-analysis for the outcomes of mean intracranial pressure, blood pressure, body temperature or adverse events. However, data from three studies showed no difference in mean intracranial pressure between the groups. Data from another study showed no evidence of a difference in blood pressure or body temperature between the progesterone and placebo groups, although there was evidence that intravenous progesterone infusion increased the frequency of phlebitis (882 participants). There was no evidence of a difference in the rate of other adverse events between progesterone treatment and placebo in the other three studies that reported on adverse events. AUTHORS' CONCLUSIONS This updated review did not find evidence that progesterone could reduce mortality or disability in patients with TBI. However, concerns regarding inconsistency (heterogeneity among participants and the intervention used) across included studies reduce our confidence in these results.There is no evidence from the available data that progesterone therapy results in more adverse events than placebo, aside from evidence from a single study of an increase in phlebitis (in the case of intravascular progesterone).There were not enough data on the effects of progesterone therapy for our other outcomes of interest (intracranial pressure, blood pressure, body temperature) for us to be able to draw firm conclusions.Future trials would benefit from a more precise classification of TBI and attempts to optimise progesterone dosage and scheduling.
Collapse
Affiliation(s)
- Junpeng Ma
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Siqing Huang
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Shu Qin
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Chao You
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Yunhui Zeng
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | | |
Collapse
|
93
|
Georgakis MK, Kalogirou EI, Diamantaras AA, Daskalopoulou SS, Munro CA, Lyketsos CG, Skalkidou A, Petridou ET. Age at menopause and duration of reproductive period in association with dementia and cognitive function: A systematic review and meta-analysis. Psychoneuroendocrinology 2016; 73:224-243. [PMID: 27543884 DOI: 10.1016/j.psyneuen.2016.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 07/27/2016] [Accepted: 08/02/2016] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The preponderance of dementia among postmenopausal women compared with same-age men and the female sex hormones neuroprotective properties support a tentative role of their deficiency in the dementia pathogenesis. METHODS Pairs of independent reviewers screened 12,323 publications derived from a search strategy for MEDLINE to identify articles investigating the association of age at menopause/reproductive period with (i) dementia and (ii) cognitive function; a snowball of eligible articles and reviews was conducted and authors were contacted for additional information. Random-effect models were used for the meta-analysis. RESULTS Age at menopause (13 studies; 19,449 participants) and reproductive period (4 studies; 9916 participants) in the highest categories were not associated with odds of dementia (effect size [ES]: 0.97 [0.78-1.21]) and Alzheimer's disease (ES: 1.06 [0.71-1.58]). Significant heterogeneity was however noted in both analyses (I2: 63.3%, p=0.003 and I2: 72.6%, p=0.01, respectively). Subgroup analyses by outcome assessment, study design, level of adjustment and study quality did not materially change the findings. In 9/13 studies assessing cognitive function, advanced age at menopause/longer reproductive period was significantly associated with better cognitive performance/lower decline. Due to statistical differences, no meta-analysis was possible for cognitive function. CONCLUSIONS Existing evidence does not support an association between indices of prolonged exposure to female hormones and lower dementia risk. There are indications, however, for better cognitive performance and delayed cognitive decline, supporting a link between female hormone deficiency and cognitive aging. Current literature limitations, indicated by the heterogeneous study-set, point towards research priorities in this clinically relevant area.
Collapse
Affiliation(s)
- Marios K Georgakis
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| | - Eleni I Kalogirou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| | - Andreas-Antonios Diamantaras
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece; Program Medical Neurosciences, Charité-Universitätsmedizin, Berlin, 10117, Germany.
| | - Stella S Daskalopoulou
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, H3G 1A4, Canada.
| | - Cynthia A Munro
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| | - Constantine G Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| | - Alkistis Skalkidou
- Department of Women's and Children's Health, Uppsala University, Uppsala, 751 85, Sweden.
| | - Eleni Th Petridou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| |
Collapse
|
94
|
van Driel CMG, Oosterwijk JC, Meijers-Heijboer EJ, van Asperen CJ, Zeijlmans van Emmichoven IA, de Vries J, Mourits MJE, Henneman L, Timmermans DRM, de Bock GH. Psychological factors associated with the intention to choose for risk-reducing mastectomy in family cancer clinic attendees. Breast 2016; 30:66-72. [PMID: 27639031 DOI: 10.1016/j.breast.2016.08.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Women seeking counseling because of familial breast cancer occurrence face difficult decisions, such as whether and when to opt for risk-reducing mastectomy (RRM) in case of BRCA1/2 mutation. Only limited research has been done to identify the psychological factors associated with the decision for RRM. This study investigated which psychological factors are related to the intention to choose for RRM. MATERIALS & METHODS A cohort of 486 cancer-unaffected women with a family history of breast cancer completed the following questionnaires prior to genetic counseling: the Cancer Worry Scale, Positive And Negative Affect Scale, Perceived Personal Control Scale, Hospital Anxiety and Depression Scale and State Anxiety Scale and questions regarding socio-demographic characteristics, family history, risk perception and RRM intention. Multivariate logistic regression was used to analyze the relation between psychological factors and women's intention to choose for RRM. RESULTS Factors associated with RRM intention were high positive affect (OR = 1.86, 95%CI = 1.12-3.08), high negative affect (OR = 2.52, 95%CI = 1.44-4.43), high cancer worry (OR = 1.65, 95%CI = 1.00-2.72), high perceived personal control (OR = 3.58, 95%CI = 2.18-5.89), high risk-perception (OR = 1.85, 95%CI = 1.15-2.95) and having children (OR = 2.06, 95%CI = 1.21-3.50). CONCLUSION Negative and positive affects play an important role in the intention for RRM. Furthermore, perceived personal control over the situation is associated with an intention for RRM. In addition to focusing on accurate risk communication, counseling should pay attention to the influence of perceived control and emotions to facilitate decision-making.
Collapse
Affiliation(s)
- C M G van Driel
- Department of Obstetrics & Gynecology, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands.
| | - J C Oosterwijk
- Department of Genetics, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - E J Meijers-Heijboer
- Department of Clinical Genetics, VU University Medical Center Amsterdam, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - C J van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - I A Zeijlmans van Emmichoven
- Department of Medical Psychology, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - J de Vries
- Department of Surgery, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - M J E Mourits
- Department of Obstetrics & Gynecology, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - L Henneman
- Department of Clinical Genetics, VU University Medical Center Amsterdam, PO Box 7057, 1007 MB Amsterdam, The Netherlands; EMGO Institute for Health and Care Research, VU University Medical Center Amsterdam, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - D R M Timmermans
- EMGO Institute for Health and Care Research, VU University Medical Center Amsterdam, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Department of Public and Occupational Health, VU University Medical Center Amsterdam, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - G H de Bock
- Department of Epidemiology, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
95
|
Giatti S, Melcangi RC, Pesaresi M. The other side of progestins: effects in the brain. J Mol Endocrinol 2016; 57:R109-26. [PMID: 27339142 DOI: 10.1530/jme-16-0061] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/22/2016] [Indexed: 01/06/2023]
Abstract
Progestins are a broad class of progestational agents widely differing in their chemical structures and pharmacological properties. Despite emerging data suggest that progestins, besides their action as endometrial protection, can also have multiple nonreproductive functions, much remains to be discovered regarding the actions exerted by these molecules in the nervous system. Here, we report the role exerted by different progestins, currently used for contraception or in postmenopausal hormone replacement therapies, in regulating cognitive functions as well as social behavior and mood. We provide evidence that the effects and mechanisms underlying their actions are still confusing due to the use of different estrogens and progestins as well as different doses, duration of exposure, route of administration, baseline hormonal status and age of treated women. We also discuss the emerging issue concerning the relevant increase of these substances in the environment, able to deeply affect aquatic wildlife as well as to exert a possible influence in humans, which may be exposed to these compounds via contaminated drinking water and seafood. Finally, we report literature data showing the neurobiological action of progestins and in particular their importance during neurodegenerative events. This is extremely interesting, since some of the progestins currently used in clinical practice exert neuroprotective and anti-inflammatory effects in the nervous system, opening new promising opportunities for the use of these molecules as therapeutic agents for trauma and neurodegenerative disorders.
Collapse
Affiliation(s)
- Silvia Giatti
- Department of Pharmacological and Biomolecular SciencesCenter of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular SciencesCenter of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Marzia Pesaresi
- Department of Pharmacological and Biomolecular SciencesCenter of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
96
|
Tskitishvili E, Pequeux C, Munaut C, Viellevoye R, Nisolle M, Noël A, Foidart JM. Use of estetrol with other steroids for attenuation of neonatal hypoxic-ischemic brain injury: to combine or not to combine? Oncotarget 2016; 7:33722-43. [PMID: 27231853 PMCID: PMC5085115 DOI: 10.18632/oncotarget.9591] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Estetrol (E4), estradiol (E2) and progesterone (P4) have important antioxidative and neuroprotective effects in neuronal system. We aimed to study the consequence of combined steroid therapy in neonatal hypoxic-ischemic encephalopathy (HIE). In vitro the effect of E4 combined with other steroids on oxidative stress and the cell viability in primary hippocampal cultures was evaluated by lactate dehydrogenase and cell survival assays. In vivo neuroprotective and therapeutic efficacy of E4 combined with other steroids was studied in HIE model of immature rats. The rat pups rectal temperature, body and brain weights were evaluated.The hippocampus and the cortex were investigated by histo/immunohistochemistry: intact cell number counting, expressions of markers for early gray matter lose, neuro- and angiogenesis were studied. Glial fibrillary acidic protein was evaluated by ELISA in blood samples. In vitro E4 and combinations of high doses of E4 with P4 and/or E2 significantly diminished the LDH activity and upregulated the cell survival.In vivopretreatment or treatment by different combinations of E4 with other steroids had unalike effects on body and brain weight, neuro- and angiogenesis, and GFAP expression in blood. The combined use of E4 with other steroids has no benefit over the single use of E4.
Collapse
Affiliation(s)
- Ekaterine Tskitishvili
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Christel Pequeux
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Carine Munaut
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Renaud Viellevoye
- Neonatal Intensive Care Unit, Department of Pediatrics, University of Liege, Liege 1, Belgium
| | - Michelle Nisolle
- Department of Obstetrics and Gynecology, University of Liege, Liege1, Belgium
| | - Agnes Noël
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| | - Jean-Michel Foidart
- Laboratory of Development Biology and Tumor, GIGA-Cancer, Department of Obstetrics and Gynecology/Department of Clinical Sciences, University of Liege, Liege 1, Belgium
| |
Collapse
|
97
|
Vaňková M, Hill M, Velíková M, Včelák J, Vacínová G, Dvořáková K, Lukášová P, Vejražková D, Rusina R, Holmerová I, Jarolímová E, Vaňková H, Kancheva R, Bendlová B, Stárka L. Preliminary evidence of altered steroidogenesis in women with Alzheimer's disease: Have the patients "OLDER" adrenal zona reticularis? J Steroid Biochem Mol Biol 2016; 158:157-177. [PMID: 26704533 DOI: 10.1016/j.jsbmb.2015.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) represents more than half of total dementias. Various factors including altered steroid biosynthesis may participate in its pathophysiology. We investigated how the circulating steroids (measured by GC-MS and RIA) may be altered in the presence of AD. Sixteen women with AD and 22 age- and BMI-corresponding controls aged over 65 years were enrolled in the study. The steroid levels (47 steroids and steroid polar conjugates) and their ratios in AD female patients indicated increased CYP11A1 activity, weakened activity of the CYP17A1C17,20 lyase metabolic step and attenuated sulfotransferase SULT2A1 activity at higher activity of the CYP17A1 17-hydroxylase step. The patients showed diminished HSD3B2 activity for C21 steroids, abated conversion of 17-hydroxyprogesterone to cortisol, and significantly elevated cortisol. The women with AD had also attenuated steroid 7α-hydroxylation forming immunoprotective Δ(5)-C19 steroids, attenuated aromatase activity forming estradiol that induces autoimmunity and a shift from the 3β-hydroxy-5α/β-reduced C19 steroids to their neuroinhibitory and antiinflammatory GABAergic 3α-hydroxy- counterparts and showed higher levels of the 3α-hydroxy-5α/β-reduced C21 steroids and pregnenolone sulfate (improves cognitive abilities but may be both protective and excitotoxic). Our preliminary data indicated functioning of alternative "backdoor" pathway in women with AD showing higher levels of both 5α/β-reduced C21 steroids but reduced levels of both 5α/β-reduced C21 steroids, which implied that the alternative "backdoor" pathway might include both 5α- and 5β-reduced steroids. Our study suggested relationships between AD status in women based on the age of subjects and levels of 10 steroids measured by GC-MS.
Collapse
Affiliation(s)
- Markéta Vaňková
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Martin Hill
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Marta Velíková
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Josef Včelák
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Gabriela Vacínová
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | | | - Petra Lukášová
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | | | - Robert Rusina
- Department of Neurology, Thomayer's Hospital, Vídeňská 800, Prague 140 59, Czech Republic.
| | - Iva Holmerová
- Faculty of Humanities, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic.
| | - Eva Jarolímová
- Faculty of Humanities, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic.
| | - Hana Vaňková
- Faculty of Humanities, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic; Third Faculty of Medicine, Charles University in Prague, Ovocný trh 5, Prague 110 00, Czech Republic.
| | - Radmila Kancheva
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Běla Bendlová
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| | - Luboslav Stárka
- Institute of Endocrinology, Národní 8, Prague 116 94, Czech Republic.
| |
Collapse
|
98
|
Jiang C, Zuo F, Wang Y, Wan J, Yang Z, Lu H, Chen W, Zang W, Yang Q, Wang J. Progesterone exerts neuroprotective effects and improves long-term neurologic outcome after intracerebral hemorrhage in middle-aged mice. Neurobiol Aging 2016; 42:13-24. [PMID: 27143417 DOI: 10.1016/j.neurobiolaging.2016.02.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/27/2016] [Accepted: 02/28/2016] [Indexed: 11/26/2022]
Abstract
In this study, we examined the effect of progesterone on histopathologic and functional outcomes of intracerebral hemorrhage (ICH) in 10- to 12-month-old mice. Progesterone or vehicle was administered by intraperitoneal injection 1 hour after collagenase-induced ICH and then by subcutaneous injections at 6, 24, and 48 hours. Oxidative and nitrosative stress were assayed at 12 hours post-ICH. Injury markers were examined on day 1, and lesion was examined on day 3. Neurologic deficits were examined for 28 days. Progesterone posttreatment reduced lesion volume, brain swelling, edema, and cell degeneration and improved long-term neurologic function. These protective effects were associated with reductions in protein carbonyl formation, protein nitrosylation, and matrix metalloproteinase-9 activity and attenuated cellular and molecular inflammatory responses. Progesterone also reduced vascular endothelial growth factor expression, increased neuronal-specific Na(+)/K(+) ATPase ɑ3 subunit expression, and reduced protein kinase C-dependent Na(+)/K(+) ATPase phosphorylation. Furthermore, progesterone reduced glial scar thickness, myelin loss, brain atrophy, and residual injury volume on day 28 after ICH. With multiple brain targets, progesterone warrants further investigation for its potential use in ICH therapy.
Collapse
Affiliation(s)
- Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China; Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | - Fangfang Zuo
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| | - Yuejuan Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| | - Jieru Wan
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Zengjin Yang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Wenwu Chen
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Weidong Zang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Qingwu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Jian Wang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
99
|
Contribution of sex hormones to gender differences in schizophrenia: A review. Asian J Psychiatr 2015; 18:2-14. [PMID: 26321672 DOI: 10.1016/j.ajp.2015.07.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/14/2015] [Accepted: 07/25/2015] [Indexed: 12/22/2022]
Abstract
Female patients with schizophrenia tend to have a more benign course and better outcomes than males. One proposed explanation is the differential influence of male and female sex hormones, including estrogen, progesterone, testosterone, and dehydroepiandrosterone (DHEA) and its sulfate (DHEAS). Such benefit may be mediated by their effects on neurotransmitters and neuroprotection. Besides altered estrogen and DHEA/DHEAS levels in female patients, data is equivocal on hormonal differences between patients and controls. However, several reports note a mostly negative correlation between estrogen levels and symptom severity in both genders, and a positive correlation between estrogen levels and neurocognition but mainly in females. Adjunctive estrogen appears to improve symptoms in both genders. Progesterone levels have inconsistent links to symptom severity in both genders, and correlate positively with neurocognition but only in males. Estrogen-progesterone combination shows preliminary benefits as augmentation for both symptoms and neurocognition in females. Testosterone levels correlate inversely with negative symptoms in males and have inconsistent associations with neurocognition in both genders. Testosterone augmentation reduced negative symptoms in male patients in a pilot investigation, but has not been evaluated for neurocognition in either gender. DHEA/DHEAS have mixed results for their association with, and clinical utility for, symptoms and neurocognition in both genders. Overall, data on the impact of sex hormones on clinical course or as treatment for schizophrenia is limited, but estrogen has most evidence for positive influence and clinical benefit. The possibly greater tolerability and broader impact of these hormones versus existing medications support further exploration of their use.
Collapse
|
100
|
Stanojlović M, Guševac I, Grković I, Zlatković J, Mitrović N, Zarić M, Horvat A, Drakulić D. Effects of chronic cerebral hypoperfusion and low-dose progesterone treatment on apoptotic processes, expression and subcellular localization of key elements within Akt and Erk signaling pathways in rat hippocampus. Neuroscience 2015; 311:308-21. [DOI: 10.1016/j.neuroscience.2015.10.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 10/19/2015] [Accepted: 10/21/2015] [Indexed: 12/12/2022]
|